File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: HN1L/AP-2γ/PLK1 signaling drives tumor progression and chemotherapy resistance in esophageal squamous cell carcinoma

TitleHN1L/AP-2γ/PLK1 signaling drives tumor progression and chemotherapy resistance in esophageal squamous cell carcinoma
Authors
Issue Date1-Dec-2022
Citation
Cell Death & Disease, 2022, v. 13 How to Cite?
Abstract

Hematological and neurological expressed 1 like (HN1L) is a newly identified oncogene in lung cancer and hepatocellular carcinoma recently identified by our team, but its roles in the development and treatment of esophageal squamous cell carcinoma (ESCC) remain incompletely cataloged. Here, using ESCC tissue array and public database analysis, we demonstrated that HN1L was highly expressed in ESCC tissues, which was associated with tumor tissue invasion, poor clinical stage and short survival for ESCC patients. Loss- and gain-of-function studies in ESCC cells revealed that HN1L enhances ESCC cell metastasis and proliferation in vitro and in mice models. Moreover, high level of HN1L reduces the sensibility of ESCC cells to chemotherapeutic drugs, such as Docetaxel. Mechanism studies revealed that HN1L activated the transcription of polo-like kinase 1 (PLK1) by interacting with transcription factor AP-2γ, which increased the expression of malignancy related proteins Cyclin D1 and Slug in ESCC cells. Blocking PLK1 with inhibitor BI-2356 abrogated the oncogenic function of HN1L and significantly suppressed ESCC progression by combining with chemotherapy. Therefore, this study demonstrates the vital pro-tumor role of HN1L/AP-2γ/PLK1 signaling axis in ESCC, offering a potential therapeutic strategy for ESCC patients with high HN1L by blocking PLK1.


Persistent Identifierhttp://hdl.handle.net/10722/328370

 

DC FieldValueLanguage
dc.contributor.authorZeng, TT-
dc.contributor.authorDeng, TH-
dc.contributor.authorLiu, Z-
dc.contributor.authorZhan, JR-
dc.contributor.authorMa, YZ-
dc.contributor.authorYan, YY-
dc.contributor.authorSun, X-
dc.contributor.authorZhu, YH-
dc.contributor.authorLi, Y-
dc.contributor.authorGuan, XY-
dc.contributor.authorLi, L-
dc.date.accessioned2023-06-28T04:43:32Z-
dc.date.available2023-06-28T04:43:32Z-
dc.date.issued2022-12-01-
dc.identifier.citationCell Death & Disease, 2022, v. 13-
dc.identifier.urihttp://hdl.handle.net/10722/328370-
dc.description.abstract<p>Hematological and neurological expressed 1 like (HN1L) is a newly identified oncogene in lung cancer and hepatocellular carcinoma recently identified by our team, but its roles in the development and treatment of esophageal squamous cell carcinoma (ESCC) remain incompletely cataloged. Here, using ESCC tissue array and public database analysis, we demonstrated that HN1L was highly expressed in ESCC tissues, which was associated with tumor tissue invasion, poor clinical stage and short survival for ESCC patients. Loss- and gain-of-function studies in ESCC cells revealed that HN1L enhances ESCC cell metastasis and proliferation in vitro and in mice models. Moreover, high level of HN1L reduces the sensibility of ESCC cells to chemotherapeutic drugs, such as Docetaxel. Mechanism studies revealed that HN1L activated the transcription of polo-like kinase 1 (PLK1) by interacting with transcription factor AP-2γ, which increased the expression of malignancy related proteins Cyclin D1 and Slug in ESCC cells. Blocking PLK1 with inhibitor BI-2356 abrogated the oncogenic function of HN1L and significantly suppressed ESCC progression by combining with chemotherapy. Therefore, this study demonstrates the vital pro-tumor role of HN1L/AP-2γ/PLK1 signaling axis in ESCC, offering a potential therapeutic strategy for ESCC patients with high HN1L by blocking PLK1.<br></p>-
dc.languageeng-
dc.relation.ispartofCell Death & Disease-
dc.titleHN1L/AP-2γ/PLK1 signaling drives tumor progression and chemotherapy resistance in esophageal squamous cell carcinoma-
dc.typeArticle-
dc.identifier.doi10.1038/s41419-022-05478-1-
dc.identifier.hkuros344666-
dc.identifier.volume13-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats